1
|
Zhang S, Geng Y, Jiang X, Sun Z, Yan M, Bi J, Tian X, Wang Q. Investigating the mechanisms of inflammation and immune alterations in Parkinson's disease using spatial transcriptomics techniques. Brain Res Bull 2024; 217:111076. [PMID: 39306046 DOI: 10.1016/j.brainresbull.2024.111076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024]
Abstract
In recent years, overwhelming evidence has emphasized the crucial role of inflammation in the pathogenesis of PD. However, the exact mechanisms by which inflammation damages dopaminergic neurons in PD are still unclear. Therefore, we generated a MPTP-induced PD mouse model and performed spatial transcriptomic sequencing to provide more insight into the process of PD development at specific brain regions. Our results indicate that the pathological changes of PD are mainly manifested in the midbrain, especially in the substantia nigra region, with significant reductions in oligodendrocytes and Agt-labeled astrocytes and an increase in Gfap-labeled astrocytes. Macrophages displayed an increasing trend in the PD environment, indicating a pattern of immune modulation induced by PD. Moreover, pathway analysis revealed significant impairments in ion migration ability, abnormal Ca2+ channels, cAMP signaling, and synaptic damage in PD. Significant downregulation of Mt1 and Mt2 and upregulation of Atp1b2, Gpi1, and Cox6a1 in PD further underscored the occurrence of intense inflammation and immune alterations. On the basis of these findings, we have validated the significant accumulation of Ca2+ in the midbrain tissue in the PD environment by measuring its content. Additionally, we have demonstrated a close association between the reduction of dopaminergic neurons, represented by the midbrain region, and ferroptosis by evaluating the iron content, malondialdehyde (MDA) levels, and the protein expression of GPX4 and TH in the tissue. We propose the hypothesis that PD-related inflammation and immune changes can induce neuronal and oligodendrocyte damage through the induction of ferroptosis, thereby further accelerating the progression of PD.
Collapse
Affiliation(s)
- Sen Zhang
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China
| | - Yifan Geng
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xing Jiang
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China
| | - Zhiyuan Sun
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China
| | - Min Yan
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China
| | - Jun Bi
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China
| | - Xuewen Tian
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China.
| | - Qinglu Wang
- Graduate School of Education, Shandong Sport University, Jinan, Shandong 250102, China.
| |
Collapse
|
2
|
Bettinger CM, Dulz S, Atiskova Y, Guerreiro H, Schön G, Guder P, Maier SL, Denecke J, Bley AE. Overview of Neuro-Ophthalmic Findings in Leukodystrophies. J Clin Med 2024; 13:5114. [PMID: 39274327 PMCID: PMC11396446 DOI: 10.3390/jcm13175114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/16/2024] Open
Abstract
Background: Leukodystrophies are a group of rare genetic diseases that primarily affect the white matter of the central nervous system. The broad spectrum of metabolic and pathological causes leads to manifestations at any age, most often in childhood and adolescence, and a variety of symptoms. Leukodystrophies are usually progressive, resulting in severe disabilities and premature death. Progressive visual impairment is a common symptom. Currently, no overview of the manifold neuro-ophthalmologic manifestations and visual impact of leukodystrophies exists. Methods: Data from 217 patients in the Hamburg leukodystrophy cohort were analyzed retrospectively for neuro-ophthalmologic manifestations, age of disease onset, and magnetic resonance imaging, visual evoked potential, and optical coherence tomography findings and were compared with data from the literature. Results: In total, 68% of the patients suffered from neuro-ophthalmologic symptoms, such as optic atrophy, visual neglect, strabismus, and nystagmus. Depending on the type of leukodystrophy, neuro-ophthalmologic symptoms occurred early or late during the course of the disease. Magnetic resonance imaging scans revealed pathologic alterations in the visual tract that were temporally correlated with symptoms. Conclusions: The first optical coherence tomography findings in Krabbe disease and metachromatic leukodystrophy allow retinal assessments. Comprehensive literature research supports the results of this first overview of neuro-ophthalmologic findings in leukodystrophies.
Collapse
Affiliation(s)
| | - Simon Dulz
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yevgeniya Atiskova
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Helena Guerreiro
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Gerhard Schön
- Center of Experimental Medicine, Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Philipp Guder
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sarah Lena Maier
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jonas Denecke
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Annette E Bley
- Children's Hospital, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
3
|
Mura E, Parazzini C, Tonduti D. Rare forms of hypomyelination and delayed myelination. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:225-252. [PMID: 39322381 DOI: 10.1016/b978-0-323-99209-1.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Hypomyelination is defined by the evidence of an unchanged pattern of deficient myelination on two MRIs performed at least 6 months apart in a child older than 1 year. When the temporal criteria are not fulfilled, and the follow-up MRI shows a progression of the myelination even if still not adequate for age, hypomyelination is excluded and the pattern is instead consistent with delayed myelination. This can be mild and nonspecific in some cases, while in other cases there is a severe delay that in the first disease stages could be difficult to differentiate from hypomyelination. In hypomyelinating leukodystrophies, hypomyelination is due to a primary impairment of myelin deposition, such as in Pelizaeus Merzabcher disease. Conversely, myelin lack is secondary, often to primary neuronal disorders, in delayed myelination and some condition with hypomyelination. Overall, the group of inherited white matter disorders with abnormal myelination has expanded significantly during the past 20 years. Many of these disorders have only recently been described, for many of them only a few patients have been reported and this contributes to make challenging the diagnostic process and the interpretation of Next Generation Sequencing results. In this chapter, we review the clinical and radiologic features of rare and lesser known forms of hypomyelination and delayed myelination not mentioned in other chapters of this handbook.
Collapse
Affiliation(s)
- Eleonora Mura
- Unit of Pediatric Neurology, Department of Biomedical and Clinical Sciences, V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy
| | - Cecilia Parazzini
- C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; Pediatric Radiology and Neuroradiology Department, V. Buzzi Children's Hospital, Milan, Italy
| | - Davide Tonduti
- Unit of Pediatric Neurology, Department of Biomedical and Clinical Sciences, V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Simeon R, Berardi A, Valente D, Volpi T, Vagni S, Galeoto G. Occupational Therapy Intervention in the Child with Leukodystrophy: Case Report. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1257. [PMID: 37508754 PMCID: PMC10377904 DOI: 10.3390/children10071257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND There are many different types of Leukodystrophies. Specifically, children with hypomyelination and congenital cataract syndrome (HCC) in addition to motor retardation development, hypotonia and progressive spastic paraplegia, associated with cerebellar ataxia and peripheral neuropathy, have early bilateral cataracts and intellectual disability as pathognomonic symptoms. HCC rehabilitation treatment is not well defined, but a significant amount of evidence in the literature has demonstrated the effectiveness of occupational therapy (OT) treatment in children with similar symptomatology. For this reason, the aim of this study was to describe the improvement in the autonomies and social participation of a child with HCC following OT treatment. METHODS A.E. was a 9-year-old child with HCC with severe intellectual disability. OT intervention lasted 3 months biweekly and each session lasted 45 min. Each session was divided into two parts: The first part aimed to increase the child's active involvement through activities; the second part involved training in Activities of Daily living (ADL). The outcome measures were: ABILHAND-Kids; Pediatric Evaluation of Disability Inventory; Comprehensive OT Evaluation Scale; ADL and Instrumental Activities of Daily Living. RESULTS A.E.'s outcome measure reported an improvement from an autonomy standpoint and in the child's general activity participation; there was also an increase in A.E.'s interpersonal skills. CONCLUSION OT treatment improved A.E.'s autonomy.
Collapse
Affiliation(s)
- Rachele Simeon
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Anna Berardi
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- IRCSS Neuromed, Via Atinense, 18, 86077 Pozzilli, Italy
| | - Donatella Valente
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- IRCSS Neuromed, Via Atinense, 18, 86077 Pozzilli, Italy
| | | | - Samuele Vagni
- School of Occupational Therapy, Sapienza University of Rome, 00185 Rome, Italy
| | - Giovanni Galeoto
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy
- IRCSS Neuromed, Via Atinense, 18, 86077 Pozzilli, Italy
| |
Collapse
|
5
|
NKX6-2 Disease in Two Unrelated Patients with Early Onset Spastic Quadriplegia and Diffuse Hypomyelinating Leukodystrophy. BRAIN DISORDERS 2023. [DOI: 10.1016/j.dscb.2023.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
|
6
|
Jamann H, Cui QL, Desu HL, Pernin F, Tastet O, Halaweh A, Farzam-kia N, Mamane VH, Ouédraogo O, Cleret-Buhot A, Daigneault A, Balthazard R, Klement W, Lemaître F, Arbour N, Antel J, Stratton JA, Larochelle C. Contact-Dependent Granzyme B-Mediated Cytotoxicity of Th17-Polarized Cells Toward Human Oligodendrocytes. Front Immunol 2022; 13:850616. [PMID: 35479072 PMCID: PMC9035748 DOI: 10.3389/fimmu.2022.850616] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is characterized by the loss of myelin and of myelin-producing oligodendrocytes (OLs) in the central nervous system (CNS). Pro-inflammatory CD4+ Th17 cells are considered pathogenic in MS and are harmful to OLs. We investigated the mechanisms driving human CD4+ T cell-mediated OL cell death. Using fluorescent and brightfield in vitro live imaging, we found that compared to Th2-polarized cells, Th17-polarized cells show greater interactions with primary human OLs and human oligodendrocytic cell line MO3.13, displaying longer duration of contact, lower mean speed, and higher rate of vesicle-like structure formation at the sites of contact. Using single-cell RNA sequencing, we assessed the transcriptomic profile of primary human OLs and Th17-polarized cells in direct contact or separated by an insert. We showed that upon close interaction, OLs upregulate the expression of mRNA coding for chemokines and antioxidant/anti-apoptotic molecules, while Th17-polarized cells upregulate the expression of mRNA coding for chemokines and pro-inflammatory cytokines such as IL-17A, IFN-γ, and granzyme B. We found that secretion of CCL3, CXCL10, IFN-γ, TNFα, and granzyme B is induced upon direct contact in cocultures of human Th17-polarized cells with human OLs. In addition, we validated by flow cytometry and immunofluorescence that granzyme B levels are upregulated in Th17-polarized compared to Th2-polarized cells and are even higher in Th17-polarized cells upon direct contact with OLs or MO3.13 cells compared to Th17-polarized cells separated from OLs by an insert. Moreover, granzyme B is detected in OLs and MO3.13 cells following direct contact with Th17-polarized cells, suggesting the release of granzyme B from Th17-polarized cells into OLs/MO3.13 cells. To confirm granzyme B–mediated cytotoxicity toward OLs, we showed that recombinant human granzyme B can induce OLs and MO3.13 cell death. Furthermore, pretreatment of Th17-polarized cells with a reversible granzyme B blocker (Ac-IEPD-CHO) or a natural granzyme B blocker (serpina3N) improved survival of MO3.13 cells upon coculture with Th17 cells. In conclusion, we showed that human Th17-polarized cells form biologically significant contacts with human OLs and exert direct toxicity by releasing granzyme B.
Collapse
Affiliation(s)
- Hélène Jamann
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Qiao-Ling Cui
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Haritha L. Desu
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Florian Pernin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Olivier Tastet
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
| | - Alexandre Halaweh
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Microbiology, Immunology and Infectiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Negar Farzam-kia
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Victoria Hannah Mamane
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Oumarou Ouédraogo
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Microbiology, Immunology and Infectiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
| | - Audrey Daigneault
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
| | - Renaud Balthazard
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Wendy Klement
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Florent Lemaître
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Nathalie Arbour
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jack Antel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Catherine Larochelle
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, QC, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Catherine Larochelle,
| |
Collapse
|
7
|
Hypomyelinating leukodystrophy – NKX6–2 gene variant as a cause. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
8
|
Abstract
Hypomyelinating leukodystrophies constitute a subset of genetic white matter disorders characterized by a primary lack of myelin deposition. Most patients with severe hypomyelination present in infancy or early childhood and develop severe neurological deficits, but the clinical presentation can also be mild with onset of symptoms in adolescence or adulthood. MRI can be used to visualize the process of myelination in detail, and MRI pattern recognition can provide a clinical diagnosis in many patients. Next-generation sequencing provides a definitive diagnosis in 80-90% of patients. Genes associated with hypomyelination include those that encode structural myelin proteins but also many that encode proteins involved in RNA translation and some lysosomal proteins. The precise pathomechanisms remain to be elucidated. Improved understanding of the process of myelination, the metabolic axonal support functions of myelin and the proposed contribution of myelin to CNS plasticity provide possible explanations as to why almost all patients with hypomyelination experience slow clinical decline after a long phase of stability. In this Review, we provide an overview of the hypomyelinating leukodystrophies, the advances in our understanding of myelin biology and of the genes involved in these disorders, and the insights these advances have provided into their clinical presentations and evolution.
Collapse
|
9
|
Almatrafi A, Umair M, Eldardear A, Al-Luqmani M, Hashmi JA, Albalawi AM, Alfadhel M, Ramzan K, Basit S. A homozygous missense variant in the homeobox domain of the NKX6-2 results in progressive spastic ataxia type 8 associated with lower limb weakness and neurological manifestations. J Gene Med 2020; 22:e3196. [PMID: 32246862 DOI: 10.1002/jgm.3196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Progressive spastic ataxia is a heterogeneous disorder characterized by cerebellar ataxia and limb spasticity associated with other severe neurological complications. Spastic ataxia is classified into pure and complex types, inherited in both an autosomal recessive and autosomal dominant manner. It is caused by pathogenic variants in at least eight different genes, including NKX6-2 (MIM 607063) located on chromosome 10q26.3. The present study aimed to identify the genetic variant(s) underlying progressive spastic ataxia and to establish the genotype-phenotype correlation. METHODS We collected a large consanguineous family having four affected individuals segregating progressive spastic ataxia in an autosomal recessive manner. To investigate the molecular cause of the disease, genomic DNA of three affected individuals underwent whole exome sequencing. RESULTS All of the affected individuals showed progressive clinical features such as spastic ataxia, lower limb weakness and other mild neurological abnormalities. Whole exome sequencing data were analyzed using different filters. Filtering of rare and shared homozygous variants revealed a novel homozygous missense variant (c.545C>T; p.Ala182Val) in a highly conserved homeobox domain of the NKX6-2 protein. CONCLUSIONS The findings of the present study add a novel variant to the NKX6-2 mutation spectrum and provide evidence that homozygous variants in the NKX6-2 cause progressive spastic ataxia associated with other abnormalities.
Collapse
Affiliation(s)
- Ahmad Almatrafi
- Department of Biology, College of Science, Taibah University, Almadinah Almunawwarah, Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Amr Eldardear
- College of Medicine, Taibah University, Almadinah Almunawwarah, Saudi Arabia
| | - Majid Al-Luqmani
- College of Medicine, Taibah University, Almadinah Almunawwarah, Saudi Arabia
| | - Jamil A Hashmi
- Center for Genetics and Inherited Diseases, Taibah University, Medina, Saudi Arabia
| | - Alia M Albalawi
- Center for Genetics and Inherited Diseases, Taibah University, Medina, Saudi Arabia.,Department of Biology, College of Science, King Abdulaziz University Jeddah, Jeddah, Saudi Arabia
| | - Majid Alfadhel
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs (MNGHA), Riyadh, Saudi Arabia.,Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital (KASCH), King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Khushnooda Ramzan
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
10
|
Expanding the clinical and neuroimaging features of NKX6-2-related hereditary spastic ataxia type 8. Eur J Med Genet 2020; 63:103868. [PMID: 32004679 DOI: 10.1016/j.ejmg.2020.103868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/08/2020] [Accepted: 01/25/2020] [Indexed: 12/30/2022]
Abstract
Pathogenic variants in NKX6-2 gene causing autosomal recessive spastic ataxia type 8 with hypomyelinating leukodystrophy have been reported in few families around the world. In this study, we performed Whole Exome Sequencing and identified a novel missense variant, c.501C > G; p.(Phe167Leu), in two affected siblings with main manifestations of global developmental delay, motor regression, hypotonia, clonus in lower limbs and muscle bulk atrophy especially in the upper limbs, spasticity and contracture, scoliosis, hip dislocation, oculomotor apraxia, horizontal and vertical nystagmus. In addition, wrist and foot drop due to peripheral axonal neuropathy were observed in these patients as a new clinical finding and cerebellar white matter involvement in brain Magnetic Resonance Imaging (MRI) as new imaging finding. Therefore, we expanded the manifestations of NKX6-2-related disorders in this manuscript.
Collapse
|
11
|
Helman G, Lajoie BR, Crawford J, Takanohashi A, Walkiewicz M, Dolzhenko E, Gross AM, Gainullin VG, Bent SJ, Jenkinson EM, Ferdinandusse S, Waterham HR, Dorboz I, Bertini E, Miyake N, Wolf NI, Abbink TEM, Kirwin SM, Tan CM, Hobson GM, Guo L, Ikegawa S, Pizzino A, Schmidt JL, Bernard G, Schiffmann R, van der Knaap MS, Simons C, Taft RJ, Vanderver A. Genome sequencing in persistently unsolved white matter disorders. Ann Clin Transl Neurol 2020; 7:144-152. [PMID: 31912665 PMCID: PMC6952322 DOI: 10.1002/acn3.50957] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/05/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023] Open
Abstract
Genetic white matter disorders have heterogeneous etiologies and overlapping clinical presentations. We performed a study of the diagnostic efficacy of genome sequencing in 41 unsolved cases with prior exome sequencing, resolving an additional 14 from an historical cohort (n = 191). Reanalysis in the context of novel disease-associated genes and improved variant curation and annotation resolved 64% of cases. The remaining diagnoses were directly attributable to genome sequencing, including cases with small and large copy number variants (CNVs) and variants in deep intronic and technically difficult regions. Genome sequencing, in combination with other methodologies, achieved a diagnostic yield of 85% in this retrospective cohort.
Collapse
Affiliation(s)
- Guy Helman
- Murdoch Children's Research InstituteThe Royal Children's Hospital MelbourneParkville, MelbourneAustralia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | | | - Joanna Crawford
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | - Asako Takanohashi
- Division of NeurologyChildren’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Marzena Walkiewicz
- Murdoch Children's Research InstituteThe Royal Children's Hospital MelbourneParkville, MelbourneAustralia
| | | | | | | | - Stephen J. Bent
- Data61Commonwealth Scientific and Industrial Research OrganisationBrisbaneAustralia
| | - Emma M. Jenkinson
- Faculty of Biology, Medicine and HealthSchool of Biological SciencesDivision of Evolution and Genomic SciencesUniversity of ManchesterManchesterUnited Kingdom
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic DiseasesDepartment of Clinical ChemistryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Hans R. Waterham
- Laboratory Genetic Metabolic DiseasesDepartment of Clinical ChemistryAmsterdam University Medical CentersUniversity of AmsterdamAmsterdamThe Netherlands
| | - Imen Dorboz
- INSERM UMR 1141DHU PROTECTUniversité Paris Diderot‐ SorbonneParis CitéFrance
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative DisordersLaboratory of Molecular MedicineBambino Gesu' Children's HospitalRomeItaly
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù00146RomeItaly
| | - Noriko Miyake
- Department of Human GeneticsYokohama City University Graduate School of MedicineFukuura, Kanazawa‐kuYokohama236‐0004Japan
| | - Nicole I. Wolf
- Department of Child NeurologyEmma Children’s HospitalAmsterdam University Medical CentersVrije Universiteit Amsterdam and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Truus E. M. Abbink
- Department of Child NeurologyEmma Children’s HospitalAmsterdam University Medical CentersVrije Universiteit Amsterdam and Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Susan M. Kirwin
- Molecular Diagnostics LaboratoryNemours Biomedical ResearchNemours/Alfred I. duPont Hospital for ChildrenWilmingtonDelaware
| | - Christina M. Tan
- Molecular Diagnostics LaboratoryNemours Biomedical ResearchNemours/Alfred I. duPont Hospital for ChildrenWilmingtonDelaware
| | - Grace M. Hobson
- Molecular Diagnostics LaboratoryNemours Biomedical ResearchNemours/Alfred I. duPont Hospital for ChildrenWilmingtonDelaware
| | - Long Guo
- Laboratory of Bone and Joint DiseasesRIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Shiro Ikegawa
- Laboratory of Bone and Joint DiseasesRIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Amy Pizzino
- Division of NeurologyChildren’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Johanna L. Schmidt
- Division of NeurologyChildren’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Genevieve Bernard
- Departments of Neurology and Neurosurgery, Pediatrics, and Human GeneticsMcGill UniversityMontrealCanada
- Division of Medical GeneticsMontreal Children’s HospitalMcGill University Health CenterMontrealCanada
- Child Health and Human Development ProgramResearch Institute of the McGill University Health CenterMontrealCanada
| | - Raphael Schiffmann
- Institute of Metabolic DiseaseBaylor Scott & White Research InstituteDallasTexas
| | - Marjo S. van der Knaap
- Department of Child NeurologyEmma Children’s HospitalAmsterdam University Medical CentersVrije Universiteit Amsterdam and Amsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Functional GenomicsAmsterdam NeuroscienceVU UniversityAmsterdamthe Netherlands
| | - Cas Simons
- Murdoch Children's Research InstituteThe Royal Children's Hospital MelbourneParkville, MelbourneAustralia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | | | - Adeline Vanderver
- Division of NeurologyChildren’s Hospital of PhiladelphiaPhiladelphiaPennsylvania
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| |
Collapse
|
12
|
Beaudin M, Matilla-Dueñas A, Soong BW, Pedroso JL, Barsottini OG, Mitoma H, Tsuji S, Schmahmann JD, Manto M, Rouleau GA, Klein C, Dupre N. The Classification of Autosomal Recessive Cerebellar Ataxias: a Consensus Statement from the Society for Research on the Cerebellum and Ataxias Task Force. CEREBELLUM (LONDON, ENGLAND) 2019; 18:1098-1125. [PMID: 31267374 PMCID: PMC6867988 DOI: 10.1007/s12311-019-01052-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is currently no accepted classification of autosomal recessive cerebellar ataxias, a group of disorders characterized by important genetic heterogeneity and complex phenotypes. The objective of this task force was to build a consensus on the classification of autosomal recessive ataxias in order to develop a general approach to a patient presenting with ataxia, organize disorders according to clinical presentation, and define this field of research by identifying common pathogenic molecular mechanisms in these disorders. The work of this task force was based on a previously published systematic scoping review of the literature that identified autosomal recessive disorders characterized primarily by cerebellar motor dysfunction and cerebellar degeneration. The task force regrouped 12 international ataxia experts who decided on general orientation and specific issues. We identified 59 disorders that are classified as primary autosomal recessive cerebellar ataxias. For each of these disorders, we present geographical and ethnical specificities along with distinctive clinical and imagery features. These primary recessive ataxias were organized in a clinical and a pathophysiological classification, and we present a general clinical approach to the patient presenting with ataxia. We also identified a list of 48 complex multisystem disorders that are associated with ataxia and should be included in the differential diagnosis of autosomal recessive ataxias. This classification is the result of a consensus among a panel of international experts, and it promotes a unified understanding of autosomal recessive cerebellar disorders for clinicians and researchers.
Collapse
Affiliation(s)
- Marie Beaudin
- Axe Neurosciences, CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Antoni Matilla-Dueñas
- Department of Neuroscience, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona, Barcelona, Spain
| | - Bing-Weng Soong
- Department of Neurology, Shuang Ho Hospital and Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan, Republic of China
- National Yang-Ming University School of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Jose Luiz Pedroso
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Orlando G Barsottini
- Ataxia Unit, Department of Neurology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Hiroshi Mitoma
- Medical Education Promotion Center, Tokyo Medical University, Tokyo, Japan
| | - Shoji Tsuji
- The University of Tokyo, Tokyo, Japan
- International University of Health and Welfare, Chiba, Japan
| | - Jeremy D Schmahmann
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Mario Manto
- Service de Neurologie, Médiathèque Jean Jacquy, CHU-Charleroi, 6000, Charleroi, Belgium
- Service des Neurosciences, UMons, Mons, Belgium
| | | | | | - Nicolas Dupre
- Axe Neurosciences, CHU de Québec-Université Laval, Québec, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
13
|
Yan H, Helman G, Murthy SE, Ji H, Crawford J, Kubisiak T, Bent SJ, Xiao J, Taft RJ, Coombs A, Wu Y, Pop A, Li D, de Vries LS, Jiang Y, Salomons GS, van der Knaap MS, Patapoutian A, Simons C, Burmeister M, Wang J, Wolf NI. Heterozygous Variants in the Mechanosensitive Ion Channel TMEM63A Result in Transient Hypomyelination during Infancy. Am J Hum Genet 2019; 105:996-1004. [PMID: 31587869 DOI: 10.1016/j.ajhg.2019.09.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/09/2019] [Indexed: 01/05/2023] Open
Abstract
Mechanically activated (MA) ion channels convert physical forces into electrical signals. Despite the importance of this function, the involvement of mechanosensitive ion channels in human disease is poorly understood. Here we report heterozygous missense mutations in the gene encoding the MA ion channel TMEM63A that result in an infantile disorder resembling a hypomyelinating leukodystrophy. Four unrelated individuals presented with congenital nystagmus, motor delay, and deficient myelination on serial scans in infancy, prompting the diagnosis of Pelizaeus-Merzbacher (like) disease. Genomic sequencing revealed that all four individuals carry heterozygous missense variants in the pore-forming domain of TMEM63A. These variants were confirmed to have arisen de novo in three of the four individuals. While the physiological role of TMEM63A is incompletely understood, it is highly expressed in oligodendrocytes and it has recently been shown to be a MA ion channel. Using patch clamp electrophysiology, we demonstrated that each of the modeled variants result in strongly attenuated stretch-activated currents when expressed in naive cells. Unexpectedly, the clinical evolution of all four individuals has been surprisingly favorable, with substantial improvements in neurological signs and developmental progression. In the three individuals with follow-up scans after 4 years of age, the myelin deficit had almost completely resolved. Our results suggest a previously unappreciated role for mechanosensitive ion channels in myelin development.
Collapse
Affiliation(s)
- Huifang Yan
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China; Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; Joint International Research Center of Translational and Clinical Research, Beijing 100871, China
| | - Guy Helman
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Melbourne, VIC 3052, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Swetha E Murthy
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037 USA
| | - Haoran Ji
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China; Children's Hospital of Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Joanna Crawford
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Thomas Kubisiak
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stephen J Bent
- Data61, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD 4067, Australia
| | - Jiangxi Xiao
- Department of Radiology, Peking University First Hospital, Beijing 100871, China
| | | | - Adam Coombs
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037 USA
| | - Ye Wu
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China
| | - Ana Pop
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam 1081 HV, the Netherlands; Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam 1081 HV, the Netherlands
| | - Dongxiao Li
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China; Henan Provincial Key Laboratory of Children's Genetic and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Linda S de Vries
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht 3584 EA, the Netherlands; UMC Utrecht Brain Center, Utrecht 3584 CG, the Netherlands
| | - Yuwu Jiang
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China; Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing 100871, China
| | - Gajja S Salomons
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam 1081 HV, the Netherlands; Amsterdam Gastroenterology & Metabolism, Amsterdam University Medical Centers, Amsterdam 1081 HV, the Netherlands; Department of Genetic Metabolic Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam 1081 HV, the Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam 1081 HV, the Netherlands; Department of Functional Genomics, Amsterdam Neuroscience, VU University, Amsterdam 1081 HV, the Netherlands
| | - Ardem Patapoutian
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037 USA
| | - Cas Simons
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Melbourne, VIC 3052, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; Departments of Computational Medicine & Bioinformatics, Psychiatry and Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jingmin Wang
- Department of Pediatrics, Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Peking University First Hospital, Beijing 100871, China; Joint International Research Center of Translational and Clinical Research, Beijing 100871, China; Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing 100871, China
| | - Nicole I Wolf
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam 1081 HV, the Netherlands.
| |
Collapse
|
14
|
Chelban V, Alsagob M, Kloth K, Chirita-Emandi A, Vandrovcova J, Maroofian R, Davagnanam I, Bakhtiari S, AlSayed MD, Rahbeeni Z, AlZaidan H, Malintan NT, Johannsen J, Efthymiou S, Ghayoor Karimiani E, Mankad K, Al-Shahrani SA, Beiraghi Toosi M, AlShammari M, Groppa S, Haridy NA, AlQuait L, Qari A, Huma R, Salih MA, Almass R, Almutairi FB, Hamad MH, Alorainy IA, Ramzan K, Imtiaz F, Puiu M, Kruer MC, Bierhals T, Wood NW, Colak D, Houlden H, Kaya N. Genetic and phenotypic characterization of NKX6-2-related spastic ataxia and hypomyelination. Eur J Neurol 2019; 27:334-342. [PMID: 31509304 PMCID: PMC6946857 DOI: 10.1111/ene.14082] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/21/2019] [Indexed: 12/22/2022]
Abstract
Background and purpose Hypomyelinating leukodystrophies are a heterogeneous group of genetic disorders with a wide spectrum of phenotypes and a high rate of genetically unsolved cases. Bi‐allelic mutations in NKX6‐2 were recently linked to spastic ataxia 8 with hypomyelinating leukodystrophy. Methods Using a combination of homozygosity mapping, exome sequencing, and detailed clinical and neuroimaging assessment a series of new NKX6‐2 mutations in a multicentre setting is described. Then, all reported NKX6‐2 mutations and those identified in this study were combined and an in‐depth analysis of NKX6‐2‐related disease spectrum was provided. Results Eleven new cases from eight families of different ethnic backgrounds carrying compound heterozygous and homozygous pathogenic variants in NKX6‐2 were identified, evidencing a high NKX6‐2 mutation burden in the hypomyelinating leukodystrophy disease spectrum. Our data reveal a phenotype spectrum with neonatal onset, global psychomotor delay and worse prognosis at the severe end and a childhood onset with mainly motor phenotype at the milder end. The phenotypic and neuroimaging expression in NKX6‐2 is described and it is shown that phenotypes with epilepsy in the absence of overt hypomyelination and diffuse hypomyelination without seizures can occur. Conclusions NKX6‐2 mutations should be considered in patients with autosomal recessive, very early onset of nystagmus, cerebellar ataxia with hypotonia that rapidly progresses to spasticity, particularly when associated with neuroimaging signs of hypomyelination. Therefore, it is recommended that NXK6‐2 should be included in hypomyelinating leukodystrophy and spastic ataxia diagnostic panels.
Collapse
Affiliation(s)
- V Chelban
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK.,Department of Neurology and Neurosurgery, Institute of Emergency Medicine, Chisinau, Moldova
| | - M Alsagob
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - K Kloth
- Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - A Chirita-Emandi
- Genetics Department, University 'Victor Babes', Timisoara, Romania
| | - J Vandrovcova
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK
| | - R Maroofian
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - I Davagnanam
- Brain Repair and Rehabilitation, University College London Institute of Neurology, London, UK
| | - S Bakhtiari
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, Cellular and Molecular Medicine, Department of Neurology, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - M D AlSayed
- Medical Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - Z Rahbeeni
- Medical Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - H AlZaidan
- Medical Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - N T Malintan
- Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - J Johannsen
- Department of Paediatrics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - S Efthymiou
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK
| | - E Ghayoor Karimiani
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George's University of London, London, UK
| | - K Mankad
- Great Ormond Street Hospitals, London, UK
| | | | - M Beiraghi Toosi
- Department of Paediatric Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - M AlShammari
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - S Groppa
- Department of Neurology and Neurosurgery, Institute of Emergency Medicine, Chisinau, Moldova
| | - N A Haridy
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK.,Department of Neurology and Psychiatry, Assiut University Hospital, Assiut, Egypt
| | - L AlQuait
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - A Qari
- Medical Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - R Huma
- Medical Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - M A Salih
- Neurology Division, Department of Pediatrics, College of Medicine, King Saud University KFSHRC, Riyadh, Saudi Arabia
| | - R Almass
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - F B Almutairi
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - M H Hamad
- Neurology Division, Department of Pediatrics, College of Medicine, King Saud University KFSHRC, Riyadh, Saudi Arabia
| | - I A Alorainy
- Department of Radiology & Medical Imaging, College of Medicine, King Saud University KFSHRC, Riyadh, Saudi Arabia
| | - K Ramzan
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - F Imtiaz
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| | - M Puiu
- Genetics Department, University 'Victor Babes', Timisoara, Romania
| | - M C Kruer
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, Cellular and Molecular Medicine, Department of Neurology, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - T Bierhals
- Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - N W Wood
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK
| | - D Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, KFSHRC, Riyadh, Saudi Arabia
| | - H Houlden
- Department of Neuromuscular Diseases, University College London Institute of Neurology, London, UK
| | - N Kaya
- Department of Genetics, KFSHRC, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Baldi C, Bertoli-Avella AM, Al-Sannaa N, Alfadhel M, Al-Thihli K, Alameer S, Elmonairy AA, Al Shamsi AM, Abdelrahman HA, Al-Gazali L, Shawli A, Al-Hakami F, Yavuz H, Kandaswamy KK, Rolfs A, Brandau O, Bauer P. Expanding the clinical and genetic spectra of NKX6-2-related disorder. Clin Genet 2019; 93:1087-1092. [PMID: 29388673 DOI: 10.1111/cge.13221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/26/2018] [Accepted: 01/26/2018] [Indexed: 11/29/2022]
Abstract
Hypomyelinating leukodystrophies (HLDs) affect the white matter of the central nervous system and manifest as neurological disorders. They are genetically heterogeneous. Very recently, biallelic variants in NKX6-2 have been suggested to cause a novel form of autosomal recessive HLD. Using whole-exome or whole-genome sequencing, we identified the previously reported c.196delC and c.487C>G variants in NKX6-2 in 3 and 2 unrelated index cases, respectively; the novel c.608G>A variant was identified in a sixth patient. All variants were homozygous in affected family members only. Our patients share a primary diagnosis of psychomotor delay, and they show spastic quadriparesis, nystagmus and hypotonia. Seizures and dysmorphic features (observed in 2 families each) represent an addition to the phenotype, while developmental regression (observed in 3 families) appears to be a notable and previously underestimated clinical feature. Our findings extend the clinical and mutational spectra associated with this novel form of HLD. Comparative analysis of our 10 patients and the 15 reported previously did, however, not reveal clear evidence for a genotype-phenotype correlation.
Collapse
Affiliation(s)
- C Baldi
- Centogene AG, Rostock, Germany
| | | | - N Al-Sannaa
- John Hopkins Aramco Health Care, Pediatric Services, Dhahran, Saudi Arabia
| | - M Alfadhel
- King Abdullah International Medical Research Centre, King Saud Bin Abdulaziz University for Health Sciences, Division of Genetics, Department of Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - K Al-Thihli
- Department of Genetics, Sultan Qaboos University Hospital, Muscat, Oman
| | - S Alameer
- King Saud Bin Abdulaziz University for Health Sciences, Department of Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (NGHA), Jeddah, Saudi Arabia
| | | | - A M Al Shamsi
- Department of Paediatrics, Tawam Hospital, Al-Ain, United Arab Emirates
| | - H A Abdelrahman
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University Al Ain, Al-Ain, United Arab Emirates
| | - L Al-Gazali
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University Al Ain, United Arab Emirates
| | - A Shawli
- King Saud Bin Abdulaziz University for Health Sciences, Department of Pediatrics, King Abdulaziz Medical City, Ministry of National Guard Health Affairs (NGHA), Jeddah, Saudi Arabia.,Molecular Medicine Section, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - F Al-Hakami
- Molecular Medicine Section, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia.,Molecular Medicine Section, King Abdulaziz Medical City-WR, Jeddah, Saudi Arabia
| | - H Yavuz
- Centogene AG, Rostock, Germany
| | | | - A Rolfs
- Centogene AG, Rostock, Germany.,Albrecht-Kossel-Institute for Neuroregeneration, Medical University Rostock, Rostock, Germany
| | | | - P Bauer
- Centogene AG, Rostock, Germany
| |
Collapse
|
16
|
Flex E, Martinelli S, Van Dijck A, Ciolfi A, Cecchetti S, Coluzzi E, Pannone L, Andreoli C, Radio FC, Pizzi S, Carpentieri G, Bruselles A, Catanzaro G, Pedace L, Miele E, Carcarino E, Ge X, Chijiwa C, Lewis MES, Meuwissen M, Kenis S, Van der Aa N, Larson A, Brown K, Wasserstein MP, Skotko BG, Begtrup A, Person R, Karayiorgou M, Roos JL, Van Gassen KL, Koopmans M, Bijlsma EK, Santen GWE, Barge-Schaapveld DQCM, Ruivenkamp CAL, Hoffer MJV, Lalani SR, Streff H, Craigen WJ, Graham BH, van den Elzen APM, Kamphuis DJ, Õunap K, Reinson K, Pajusalu S, Wojcik MH, Viberti C, Di Gaetano C, Bertini E, Petrucci S, De Luca A, Rota R, Ferretti E, Matullo G, Dallapiccola B, Sgura A, Walkiewicz M, Kooy RF, Tartaglia M. Aberrant Function of the C-Terminal Tail of HIST1H1E Accelerates Cellular Senescence and Causes Premature Aging. Am J Hum Genet 2019; 105:493-508. [PMID: 31447100 DOI: 10.1016/j.ajhg.2019.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/10/2019] [Indexed: 02/03/2023] Open
Abstract
Histones mediate dynamic packaging of nuclear DNA in chromatin, a process that is precisely controlled to guarantee efficient compaction of the genome and proper chromosomal segregation during cell division and to accomplish DNA replication, transcription, and repair. Due to the important structural and regulatory roles played by histones, it is not surprising that histone functional dysregulation or aberrant levels of histones can have severe consequences for multiple cellular processes and ultimately might affect development or contribute to cell transformation. Recently, germline frameshift mutations involving the C-terminal tail of HIST1H1E, which is a widely expressed member of the linker histone family and facilitates higher-order chromatin folding, have been causally linked to an as-yet poorly defined syndrome that includes intellectual disability. We report that these mutations result in stable proteins that reside in the nucleus, bind to chromatin, disrupt proper compaction of DNA, and are associated with a specific methylation pattern. Cells expressing these mutant proteins have a dramatically reduced proliferation rate and competence, hardly enter into the S phase, and undergo accelerated senescence. Remarkably, clinical assessment of a relatively large cohort of subjects sharing these mutations revealed a premature aging phenotype as a previously unrecognized feature of the disorder. Our findings identify a direct link between aberrant chromatin remodeling, cellular senescence, and accelerated aging.
Collapse
Affiliation(s)
- Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Anke Van Dijck
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium; Department of Neurology, Antwerp University Hospital, Edegem, 2650 Belgium
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Serena Cecchetti
- Microscopy Area, Core Facilities, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Elisa Coluzzi
- Department of Science, University Roma Tre, Rome, 00146 Italy
| | - Luca Pannone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Cristina Andreoli
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, 00161 Italy
| | - Francesca Clementina Radio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Simone Pizzi
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Giovanna Carpentieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy; Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, 00161 Italy
| | | | - Lucia Pedace
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Evelina Miele
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Elena Carcarino
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy; Current affiliation: Cordeliers Research Centre, Inserm 1138, Sorbonne Université, Paris, 75006 France
| | - Xiaoyan Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Current affiliation: Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chieko Chijiwa
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - M E Suzanne Lewis
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6H 3N1, Canada
| | - Marije Meuwissen
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium
| | - Sandra Kenis
- Department of Neurology, Antwerp University Hospital, Edegem, 2650 Belgium
| | | | - Austin Larson
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kathleen Brown
- Section of Clinical Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Melissa P Wasserstein
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Brian G Skotko
- Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Maria Karayiorgou
- Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA
| | - J Louw Roos
- Department of Psychiatry, University of Pretoria, Weskoppies Hospital, Pretoria, 0001 South Africa
| | - Koen L Van Gassen
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 AB the Netherlands
| | - Marije Koopmans
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 AB the Netherlands
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | | | - Claudia A L Ruivenkamp
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Mariette J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2300 RC the Netherlands
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haley Streff
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brett H Graham
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Daan J Kamphuis
- Departement of Neurology, Reinier de Graaf Ziekenhuis, Delft, 2600 GA the Netherlands
| | - Katrin Õunap
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia
| | - Karit Reinson
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, 50406 Estonia; Institute of Clinical Medicine, University of Tartu, Tartu, 50406 Estonia; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Monica H Wojcik
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clara Viberti
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Cornelia Di Gaetano
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Enrico Bertini
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Simona Petrucci
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, 00189 Italy; Division of Medical Genetics, Casa Sollievo della Sofferenza Hospital, IRCCS, San Giovanni Rotondo, 71013 Italy
| | - Alessandro De Luca
- Division of Medical Genetics, Casa Sollievo della Sofferenza Hospital, IRCCS, San Giovanni Rotondo, 71013 Italy
| | - Rossella Rota
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146 Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University, Rome, 00161 Italy; Istituto Neuromed, IRCCS, Pozzilli, 86077 Italy
| | - Giuseppe Matullo
- Department of Medical Sciences, University of Turin, Turin, 10126 Italy; Italian Institute for Genomic Medicine, Turin, 10126 Italy
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy
| | - Antonella Sgura
- Department of Science, University Roma Tre, Rome, 00146 Italy
| | - Magdalena Walkiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Current affiliation: National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Edegem, 2650 Belgium.
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, 00146 Italy.
| |
Collapse
|
17
|
Simons C, Dyment D, Bent SJ, Crawford J, D'Hooghe M, Kohlschütter A, Venkateswaran S, Helman G, Poll-The BT, Makowski CC, Ito Y, Kernohan K, Hartley T, Waisfisz Q, Taft RJ, van der Knaap MS, Wolf NI. A recurrent de novo mutation in TMEM106B causes hypomyelinating leukodystrophy. Brain 2019; 140:3105-3111. [PMID: 29186371 DOI: 10.1093/brain/awx314] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/05/2017] [Indexed: 12/28/2022] Open
Abstract
Hypomyelinating leukodystrophies are a heterogeneous group of disorders with a clinical presentation that often includes early-onset nystagmus, ataxia and spasticity and a wide range of severity. Using next-generation sequencing techniques and GeneMatcher, we identified four unrelated patients with brain hypomyelination, all with the same recurrent dominant mutation, c.754G>A p.(Asp252Asn), in TMEM106B. The mutation was confirmed as de novo in three of the cases, and the mildly affected father of the fourth affected individual was confirmed as mosaic for this variant. The protein encoded by TMEM106B is poorly characterized but is reported to have a role in regulation of lysosomal trafficking. Polymorphisms in TMEM106B are thought to modify disease onset in frontotemporal dementia, but its relation to myelination is not understood. Clinical presentation in three of the four patients is remarkably benign compared to other hypomyelinating disorders, with congenital nystagmus and mild motor delay. These findings add TMEM106B to the growing list of genes causing hypomyelinating disorders and emphasize the essential role lysosomes play in myelination.
Collapse
Affiliation(s)
- Cas Simons
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - David Dyment
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Stephen J Bent
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - Joanna Crawford
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - Marc D'Hooghe
- Department of Neurology, General Hospital Sint-Jan, Brugge, Belgium
| | - Alfried Kohlschütter
- Department of Paediatrics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Sunita Venkateswaran
- Division of Neurology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Guy Helman
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - Bwee-Tien Poll-The
- Department of Child Neurology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Yoko Ito
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Kristin Kernohan
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Taila Hartley
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Quinten Waisfisz
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Ryan J Taft
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia.,Illumina Inc, San Diego, California, USA
| | | | - Marjo S van der Knaap
- Department of Child Neurology, VU University Medical Center, and Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, VU University Medical Center, and Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Pant DC, Dorboz I, Schluter A, Fourcade S, Launay N, Joya J, Aguilera-Albesa S, Yoldi ME, Casasnovas C, Willis MJ, Ruiz M, Ville D, Lesca G, Siquier-Pernet K, Desguerre I, Yan H, Wang J, Burmeister M, Brady L, Tarnopolsky M, Cornet C, Rubbini D, Terriente J, James KN, Musaev D, Zaki MS, Patterson MC, Lanpher BC, Klee EW, Pinto E Vairo F, Wohler E, Sobreira NLDM, Cohen JS, Maroofian R, Galehdari H, Mazaheri N, Shariati G, Colleaux L, Rodriguez D, Gleeson JG, Pujades C, Fatemi A, Boespflug-Tanguy O, Pujol A. Loss of the sphingolipid desaturase DEGS1 causes hypomyelinating leukodystrophy. J Clin Invest 2019; 129:1240-1256. [PMID: 30620337 DOI: 10.1172/jci123959] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Sphingolipid imbalance is the culprit in a variety of neurological diseases, some affecting the myelin sheath. We have used whole-exome sequencing in patients with undetermined leukoencephalopathies to uncover the endoplasmic reticulum lipid desaturase DEGS1 as the causative gene in 19 patients from 13 unrelated families. Shared features among the cases include severe motor arrest, early nystagmus, dystonia, spasticity, and profound failure to thrive. MRI showed hypomyelination, thinning of the corpus callosum, and progressive thalamic and cerebellar atrophy, suggesting a critical role of DEGS1 in myelin development and maintenance. This enzyme converts dihydroceramide (DhCer) into ceramide (Cer) in the final step of the de novo biosynthesis pathway. We detected a marked increase of the substrate DhCer and DhCer/Cer ratios in patients' fibroblasts and muscle. Further, we used a knockdown approach for disease modeling in Danio rerio, followed by a preclinical test with the first-line treatment for multiple sclerosis, fingolimod (FTY720, Gilenya). The enzymatic inhibition of Cer synthase by fingolimod, 1 step prior to DEGS1 in the pathway, reduced the critical DhCer/Cer imbalance and the severe locomotor disability, increasing the number of myelinating oligodendrocytes in a zebrafish model. These proof-of-concept results pave the way to clinical translation.
Collapse
Affiliation(s)
- Devesh C Pant
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Imen Dorboz
- INSERM UMR 1141, DHU PROTECT, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Agatha Schluter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Nathalie Launay
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Javier Joya
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service, Navarrabiomed, Pamplona, Spain
| | - Maria Eugenia Yoldi
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service, Navarrabiomed, Pamplona, Spain
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.,Neuromuscular Unit, Neurology Department, Hospital Universitari de Bellvitge, c/Feixa Llarga s/n, 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mary J Willis
- Department of Pediatrics, Naval Medical Center San Diego, San Diego, California, USA
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Dorothée Ville
- Department of Neuropediatrics, Lyon University Hospital, Lyon, France
| | - Gaetan Lesca
- Department of Medical Genetics, Lyon University Hospital and GENDEV team CNRS UMR 5292, INSERM U1028, CRNL, and University Claude Bernard Lyon 1, Lyon, France
| | - Karine Siquier-Pernet
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Isabelle Desguerre
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Molecular & Behavioral Neuroscience Institute, and
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Margit Burmeister
- Molecular & Behavioral Neuroscience Institute, and.,Departments of Computational Medicine & Bioinformatics, Psychiatry and Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Lauren Brady
- Department of Pediatrics (Neuromuscular and Neurometabolics), McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Mark Tarnopolsky
- Department of Pediatrics (Neuromuscular and Neurometabolics), McMaster Children's Hospital, Hamilton, Ontario, Canada
| | | | | | | | - Kiely N James
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Damir Musaev
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Maha S Zaki
- Human Genetics and Genome Research Division, Clinical Genetics Department, National Research Centre, Cairo, Egypt
| | - Marc C Patterson
- Departments of Neurology and Pediatrics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Eric W Klee
- Department of Clinical Genomics and.,Center for Individualized Medicine, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Filippo Pinto E Vairo
- Department of Clinical Genomics and.,Center for Individualized Medicine, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth Wohler
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nara Lygia de M Sobreira
- McKusick-Nathans Institute of Genetic Medicine, and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julie S Cohen
- Moser Center for Leukodystrophies at the Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Reza Maroofian
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Hamid Galehdari
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Neda Mazaheri
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran.,Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Laurence Colleaux
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Developmental Brain Disorders Laboratory, INSERM UMR 1163, Paris, France
| | - Diana Rodriguez
- APHP, Department of Neuropediatrics, National Reference Center for Neurogenetic Disorders, Hôpital Armand-Trousseau, GHUEP, Paris, France.,GRC ConCer-LD, Sorbonne Universités, UPMC Université, Paris, France
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, California, USA
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ali Fatemi
- Moser Center for Leukodystrophies at the Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | - Odile Boespflug-Tanguy
- INSERM UMR 1141, DHU PROTECT, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Assistance Publique des Hopitaux de Paris (APHP), Reference Center for Leukodystrophies and Rare Leukoencephalopathies (LEUKOFRANCE), Hôpital Robert Debré, Paris, France
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain
| |
Collapse
|
19
|
Dewulf JP, Wiame E, Dorboz I, Elmaleh-Bergès M, Imbard A, Dumitriu D, Rak M, Bourillon A, Helaers R, Malla A, Renaldo F, Boespflug-Tanguy O, Vincent MF, Benoist JF, Wevers RA, Schlessinger A, Van Schaftingen E, Nassogne MC, Schiff M. SLC13A3 variants cause acute reversible leukoencephalopathy and α-ketoglutarate accumulation. Ann Neurol 2019; 85:385-395. [PMID: 30635937 DOI: 10.1002/ana.25412] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 01/22/2023]
Abstract
OBJECTIVE SLC13A3 encodes the plasma membrane Na+ /dicarboxylate cotransporter 3, which imports inside the cell 4 to 6 carbon dicarboxylates as well as N-acetylaspartate (NAA). SLC13A3 is mainly expressed in kidney, in astrocytes, and in the choroid plexus. We describe two unrelated patients presenting with acute, reversible (and recurrent in one) neurological deterioration during a febrile illness. Both patients exhibited a reversible leukoencephalopathy and a urinary excretion of α-ketoglutarate (αKG) that was markedly increased and persisted over time. In one patient, increased concentrations of cerebrospinal fluid NAA and dicarboxylates (including αKG) were observed. Extensive workup was unsuccessful, and a genetic cause was suspected. METHODS Whole exome sequencing (WES) was performed. Our teams were connected through GeneMatcher. RESULTS WES analysis revealed variants in SLC13A3. A homozygous missense mutation (p.Ala254Asp) was found in the first patient. The second patient was heterozygous for another missense mutation (p.Gly548Ser) and an intronic mutation affecting splicing as demonstrated by reverse transcriptase polymerase chain reaction performed in muscle tissue (c.1016 + 3A > G). Mutations and segregation were confirmed by Sanger sequencing. Functional studies performed on HEK293T cells transiently transfected with wild-type and mutant SLC13A3 indicated that the missense mutations caused a marked reduction in the capacity to transport αKG, succinate, and NAA. INTERPRETATION SLC13A3 deficiency causes acute and reversible leukoencephalopathy with marked accumulation of αKG. Urine organic acids (especially αKG and NAA) and SLC13A3 mutations should be screened in patients presenting with unexplained reversible leukoencephalopathy, for which SLC13A3 deficiency is a novel differential diagnosis. ANN NEUROL 2019;85:385-395.
Collapse
Affiliation(s)
- Joseph P Dewulf
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium.,Department of Laboratory Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Elsa Wiame
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Imen Dorboz
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Monique Elmaleh-Bergès
- Department of Pediatric Imaging, Robert Debré University Hospital, Public APHP, Paris, France
| | - Apolline Imbard
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Dana Dumitriu
- Department of Pediatric Imaging, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Malgorzata Rak
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Agnès Bourillon
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Raphaël Helaers
- Human Molecular Genetics, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Alisha Malla
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Florence Renaldo
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Leukodystrophies and Rare Leukoencephalopathies, LEUKOFRANCE, Robert Debré University Hospital, APHP, Paris, France
| | - Odile Boespflug-Tanguy
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Leukodystrophies and Rare Leukoencephalopathies, LEUKOFRANCE, Robert Debré University Hospital, APHP, Paris, France
| | - Marie-Françoise Vincent
- Department of Laboratory Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Jean-François Benoist
- Laboratory of Biochemistry, Robert Debré University Hospital, APHP, France.,Paris-Sud University, Châtenay-Malabry, France
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emile Van Schaftingen
- Laboratory of Physiological Chemistry, de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Marie-Cécile Nassogne
- Pediatric Neurology Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Manuel Schiff
- UMR1141, PROTECT, INSERM, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,Department of Pediatric Neurology and Metabolic Diseases, Robert Debré University Hospital, APHP, Paris, France.,Reference Center for Inborn Errors of Metabolism, Robert Debré University Hospital, APHP, Paris, France
| |
Collapse
|